SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Heydarkhan Hagvall Sepideh) "

Search: WFRF:(Heydarkhan Hagvall Sepideh)

  • Result 1-12 of 12
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • González-King, Hernán, et al. (author)
  • Head-to-head comparison of relevant cell sources of small extracellular vesicles for cardiac repair : Superiority of embryonic stem cells
  • 2024
  • In: Journal of Extracellular Vesicles. - : John Wiley & Sons. - 2001-3078. ; 13:5
  • Journal article (peer-reviewed)abstract
    • Small extracellular vesicles (sEV) derived from various cell sources have been demonstrated to enhance cardiac function in preclinical models of myocardial infarction (MI). The aim of this study was to compare different sources of sEV for cardiac repair and determine the most effective one, which nowadays remains limited. We comprehensively assessed the efficacy of sEV obtained from human primary bone marrow mesenchymal stromal cells (BM-MSC), human immortalized MSC (hTERT-MSC), human embryonic stem cells (ESC), ESC-derived cardiac progenitor cells (CPC), human ESC-derived cardiomyocytes (CM), and human primary ventricular cardiac fibroblasts (VCF), in in vitro models of cardiac repair. ESC-derived sEV (ESC-sEV) exhibited the best pro-angiogenic and anti-fibrotic effects in vitro. Then, we evaluated the functionality of the sEV with the most promising performances in vitro, in a murine model of MI-reperfusion injury (IRI) and analysed their RNA and protein compositions. In vivo, ESC-sEV provided the most favourable outcome after MI by reducing adverse cardiac remodelling through down-regulating fibrosis and increasing angiogenesis. Furthermore, transcriptomic, and proteomic characterizations of sEV derived from hTERT-MSC, ESC, and CPC revealed factors in ESC-sEV that potentially drove the observed functions. In conclusion, ESC-sEV holds great promise as a cell-free treatment for promoting cardiac repair following MI. 
  •  
2.
  • Helenius, Gisela, 1973, et al. (author)
  • Effect of shear stress on the expression of coagulation and fibrinolytic factors in both smooth muscle and endothelial cells in a co-culture model.
  • 2008
  • In: European surgical research. Europ?ische chirurgische Forschung. Recherches chirurgicales europ?ennes. - : S. Karger AG. - 1421-9921 .- 0014-312X. ; 40:4, s. 325-32
  • Journal article (peer-reviewed)abstract
    • Blood vessels are subjected to forces due to the flow. Endothelial cells (EC) are recipients, cross-talk with smooth muscle cells (SMC), and regulate physiology. It was hypothesized that both EC and SMC respond to shear stress, which alters the expression of factors in coagulation and fibrinolysis. METHODS: A co-culture of human saphenous vein EC (HSVEC) and human saphenous vein SMC (HSVSMC) was exposed to shear, following which the cells were separated. Gene expression of tissue factor, thrombomodulin (TM), plasminogen activator inhibitor-1 (PAI-1), tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA) were analyzed with real-time RT-PCR. Protein expression was studied with ELISA. In HSVEC, the expression of PAI-1 (x2.1), tPA (x1.8), uPA (x1.6), tissue factor (x2.5) and TM (x1.9) was upregulated after 4 h of shear compared to controls. After 24 h of shear, expression was still upregulated in tPA (x2.3) and TM (x1.6). In HSVSMC, change in expression of PAI-1 (x2.1) was present after 4 h and in uPA (x2.1), and TM (x0.4) after 24 h. Both HSVEC and HSVSMC responded to shear, which led to altered expression of coagulation and fibrinolytic factors. This indicates that SMC, and interactions between EC and SMC, are more important in the regulation of vascular wall hemostasis than earlier studies have reported.
  •  
3.
  • Helenius, Gisela, 1973, et al. (author)
  • Expression of fibrinolytic and coagulation factors in cocultured human endothelial and smooth muscle cells
  • 2004
  • In: Tissue engineering. - 1076-3279. ; 10:3-4, s. 353-60
  • Journal article (peer-reviewed)abstract
    • Interactions between endothelial cells and smooth muscle cells are interesting from a tissue-engineering point of view. We have developed a coculture system that allows direct contact between these two cell types. The fibrinolytic factors PAI-1, tPA, and uPA and the coagulation factor TF, were studied at the gene level by RT-PCR and at the protein level by ELISA. Significant changes of all studied factors were seen at the gene level in cocultured endothelial cells. tPA and TF were upregulated 4- and 7-fold, respectively, and PAI-1 and uPA were downregulated 4- and 1.5-fold, respectively, compared with single-cultured controls. In cocultured smooth muscle cells alterations of PAI-1 and TF were significant, with a 1.5-fold upregulation of PAI-1 and a 2.5-fold downregulation of TF. Results at the protein level mirrored the gene expression results. These findings indicate that cocultured endothelial cells are rendered both hypercoagulative and hyperfibrinolytic.
  •  
4.
  •  
5.
  • Heydarkhan-Hagvall, Sepideh, 1969, et al. (author)
  • DNA microarray study on gene expression profiles in co-cultured endothelial and smooth muscle cells in response to 4- and 24-h shear stress
  • 2006
  • In: Molecular and cellular biochemistry. - : Springer Science and Business Media LLC. - 0300-8177 .- 1573-4919. ; 281:1-2, s. 1-15
  • Journal article (peer-reviewed)abstract
    • Shear stress, a major hemodynamic force acting on the vessel wall, plays an important role in physiological processes such as cell growth, differentiation, remodelling, metabolism, morphology, and gene expression. We investigated the effect of shear stress on gene expression profiles in co-cultured vascular endothelial cells (ECs) and smooth muscle cells (SMCs). Human aortic ECs were cultured as a confluent monolayer on top of confluent human aortic SMCs, and the EC side of the co-culture was exposed to a laminar shear stress of 12 dyn/cm(2) for 4 or 24 h. After shearing, the ECs and SMCs were separated and RNA was extracted from the cells. The RNA samples were labelled and hybridized with cDNA array slides that contained 8694 genes. Statistical analysis showed that shear stress caused the differential expression (p < or = 0.05) of a total of 1151 genes in ECs and SMCs. In the co-cultured ECs, shear stress caused the up-regulation of 403 genes and down-regulation of 470. In the co-cultured SMCs, shear stress caused the up-regulation of 152 genes and down-regulation of 126 genes. These results provide new information on the gene expression profile and its potential functional consequences in co-cultured ECs and SMCs exposed to a physiological level of laminar shear stress. Although the effects of shear stress on gene expression in monocultured and co-cultured EC are generally similar, the response of some genes to shear stress is opposite between these two types of culture (e.g., ICAM-1 is up-regulated in monoculture and down-regulated in co-culture), which strongly indicates that EC-SMC interactions affect EC responses to shear stress.
  •  
6.
  • Heydarkhan-Hagvall, Sepideh, 1969, et al. (author)
  • Production of extracellular matrix components in tissue-engineered blood vessels
  • 2006
  • In: Tissue engineering. - : Mary Ann Liebert Inc. - 1076-3279 .- 1557-8690. ; 12:4, s. 831-42
  • Journal article (peer-reviewed)abstract
    • Morphology and compliance of tissue-engineered blood vessels (TEBV) are dependent on the culture period and production of extracellular matrix (ECM) components in order to increase the strength of the developing tissue. The aim of the present study was to evaluate the potential of TEBVs to produce an ECM similar to native arteries and veins. Human smooth muscle cells (SMC) were seeded onto the poly(glycolic acid) (PGA) scaffold and placed in bioreactors filled with DMEM supplemented with growth factors. After 6 weeks, the vessels were harvested from the bioreactors and seeded with human endothelial cells at the lumen for another 3 days. Then, the TEBVs were harvested for RNA and protein isolation for further RT-PCR and Western blot. TEBVs had a similar macroscopic appearance to that of native vessels with no visible evidence of the original PGA. Histological and immunohistochemical analyses indicated the presence of high cell density and development of a highly organized structure of ECM. After 6 weeks of culture, there were significantly lower gene expression of SMC-specific markers, such as alpha-actin, caldesmon, and vimentin, and proteoglycans, such as biglycan, decorin, and versican, and other ECM components, such as collagen I and elastin, in TEBVs, with and without pulsatile conditions, compared to that of native arteries. Gene expression of fibronectin was significantly lower in TEBVs grown during pulsatile conditions compared to that of native arteries. No difference was observed in TEBVs grown during non-pulsatile conditions. The presence of alpha-actin, collagen I, decorin, and fibronectin at protein level was demonstrated in TEBVs with and without pulsatile conditions after 6 weeks and in native veins and arteries as well. How this deviation translates into mechanical properties remains to be explored.
  •  
7.
  • Heydarkhan-Hagvall, Sepideh, 1969 (author)
  • Toward a tissue engineered blood vessel. Cell cultivation, hemodynamic influences and matrix production
  • 2005
  • Doctoral thesis (other academic/artistic)abstract
    • Cardiovascular diseases are the leading causes of mortality in the western world. Thereplacement or repair of diseased blood vessels with human autografts (vein or arteries) orsynthetic vascular grafts (ePTFE or Dacron) has become a routine treatment. Autologous graftsare not always available in all patients. Synthetic substitutes are not able to grow or remodel andare prone to complications like stenosis, thrombosis, calcification and infection. Tissueengineered blood vessels (TEBVs) could offer a promising approach to reduce the limitations insmall vessel grafting by creating viable constructs with repair and remodelling capabilities.The aim of this thesis is to form biologically active TEBVs in vitro. In particular, the importanceof a cell-cell contact in expression of angiogenic factors in the vascular wall was studied in a coculturesystem of endothelial cells (ECs) and smooth muscle cells (SMCs). Also, the effects ofthe physiological level of laminar shear stress on vascular cells in the co-culture wereinvestigated. Furthermore, the production of extracellular matrix (ECM) components in TEBVswas evaluated. Finally, mesenchymal stem cells were compared to vascular SMCs when appliedto cyclic strains in order to investigate if these cells can be an alternative cell source for vasculartissue engineering.Direct cell-cell interactions of vascular ECs and SMCs were found to affect the gene and proteinexpression of angiogenic factors. Shear stress modulated the expression profiles of genes relatedto cytoskeleton/ECM modelling, cell proliferation, signal transduction, and immuneresponses/inflammation in co-cultured ECs and SMCs. The gene expression and proteinproduction of different SMCs' specific markers and ECM components in tissue engineeredconstruct were lower in the constructs compared to native veins and arteries after a period of 6weeks. Human MSCs were demonstrated to be capable of responding to mechanical stress, andthey hold great potential as a cell source of autologous vascular cells.To increase the success of engineered vascular grafts, the blood vessel constructs should bedesigned to mimic the properties of native blood vessels. Therefore, understanding theimportance of ECM and regulation of cellular behaviour is particularly necessary.
  •  
8.
  • Johansson, Markus, et al. (author)
  • Cardiac hypertrophy in a dish: a human stem cell based mode
  • 2020
  • In: Biology Open. - : The Company of Biologists. - 2046-6390. ; 9:9
  • Journal article (peer-reviewed)abstract
    • Cardiac hypertrophy is an important and independent risk factor for the development of heart failure. To better understand the mechanisms and regulatory pathways involved in cardiac hypertrophy, there is a need for improved in vitro models. In this study, we investigated how hypertrophic stimulation affected human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs). The cells were stimulated with endothelin-1 (ET-1) for 8, 24, 48, 72, or 96 h. Parameters including cell size, ANP-, proBNP-, and lactate concentration were analyzed. Moreover, transcriptional profiling using RNA-sequencing was performed to identify differentially expressed genes following ET-1 stimulation. The results show that the CMs increase in size by approximately 13% when exposed to ET-1 in parallel to increases in ANP and proBNP protein and mRNA levels. Furthermore, the lactate concentration in the media was increased indicating that the CMs consume more glucose, a hallmark of cardiac hypertrophy. Using RNA-seq, a hypertrophic gene expression pattern was also observed in the stimulated CMs. Taken together, these results show that hiPSC-derived CMs stimulated with ET-1 display a hypertrophic response. The results from this study also provide new molecular insights about the underlying mechanisms of cardiac hypertrophy and may help accelerate the development of new drugs against this condition.
  •  
9.
  • Johansson, Markus, et al. (author)
  • Data Mining Identifies CCN2 and THBS1 as Biomarker Candidates for Cardiac Hypertrophy
  • 2022
  • In: Life-Basel. - : MDPI AG. ; 12:5
  • Journal article (peer-reviewed)abstract
    • Cardiac hypertrophy is a condition that may contribute to the development of heart failure. In this study, we compare the gene-expression patterns of our in vitro stem-cell-based cardiac hypertrophy model with the gene expression of biopsies collected from hypertrophic human hearts. Twenty-five differentially expressed genes (DEGs) from both groups were identified and the expression of selected corresponding secreted proteins were validated using ELISA and Western blot. Several biomarkers, including CCN2, THBS1, NPPA, and NPPB, were identified, which showed significant overexpressions in the hypertrophic samples in both the cardiac biopsies and in the endothelin-1-treated cells, both at gene and protein levels. The protein-interaction network analysis revealed CCN2 as a central node among the 25 overlapping DEGs, suggesting that this gene might play an important role in the development of cardiac hypertrophy. GO-enrichment analysis of the 25 DEGs revealed many biological processes associated with cardiac function and the development of cardiac hypertrophy. In conclusion, we identified important similarities between ET-1-stimulated human-stem-cell-derived cardiomyocytes and human hypertrophic cardiac tissue. Novel putative cardiac hypertrophy biomarkers were identified and validated on the protein level, lending support for further investigations to assess their potential for future clinical applications.
  •  
10.
  • Johansson, Markus, et al. (author)
  • Multi-Omics Characterization of a Human Stem Cell-Based Model of Cardiac Hypertrophy
  • 2022
  • In: Life-Basel. - : MDPI AG. - 2075-1729. ; 12:2
  • Journal article (peer-reviewed)abstract
    • Cardiac hypertrophy is an important and independent risk factor for the development of cardiac myopathy that may lead to heart failure. The mechanisms underlying the development of cardiac hypertrophy are yet not well understood. To increase the knowledge about mechanisms and regulatory pathways involved in the progression of cardiac hypertrophy, we have developed a human induced pluripotent stem cell (hiPSC)-based in vitro model of cardiac hypertrophy and performed extensive characterization using a multi-omics approach. In a series of experiments, hiPSC-derived cardiomyocytes were stimulated with Endothelin-1 for 8, 24, 48, and 72 h, and their transcriptome and secreted proteome were analyzed. The transcriptomic data show many enriched canonical pathways related to cardiac hypertrophy already at the earliest time point, e.g., cardiac hypertrophy signaling. An integrated transcriptome-secretome analysis enabled the identification of multimodal biomarkers that may prove highly relevant for monitoring early cardiac hypertrophy progression. Taken together, the results from this study demonstrate that our in vitro model displays a hypertrophic response on both transcriptomic- and secreted-proteomic levels. The results also shed novel insights into the underlying mechanisms of cardiac hypertrophy, and novel putative early cardiac hypertrophy biomarkers have been identified that warrant further investigation to assess their potential clinical relevance.
  •  
11.
  • Nawaz, Muhammad, et al. (author)
  • Lipid Nanoparticles Deliver the Therapeutic VEGFA mRNA In Vitro and In Vivo and Transform Extracellular Vesicles for Their Functional Extensions
  • 2023
  • In: Advanced Science. - : John Wiley & Sons. - 2198-3844. ; 10:12
  • Journal article (peer-reviewed)abstract
    • Lipid nanoparticles (LNPs) are currently used to transport functional mRNAs, such as COVID-19 mRNA vaccines. The delivery of angiogenic molecules, such as therapeutic VEGF-A mRNA, to ischemic tissues for producing new blood vessels is an emerging strategy for the treatment of cardiovascular diseases. Here, the authors deliver VEGF-A mRNA via LNPs and study stoichiometric quantification of their uptake kinetics and how the transport of exogenous LNP-mRNAs between cells is functionally extended by cells’ own vehicles called extracellular vesicles (EVs). The results show that cellular uptake of LNPs and their mRNA molecules occurs quickly, and that the translation of exogenously delivered mRNA begins immediately. Following the VEGF-A mRNA delivery to cells via LNPs, a fraction of internalized VEGF-A mRNA is secreted via EVs. The overexpressed VEGF-A mRNA is detected in EVs secreted from three different cell types. Additionally, RNA-Seq analysis reveals that as cells’ response to LNP-VEGF-A mRNA treatment, several overexpressed proangiogenic transcripts are packaged into EVs. EVs are further deployed to deliver VEGF-A mRNA in vitro and in vivo. Upon equal amount of VEGF-A mRNA delivery via three EV types or LNPs in vitro, EVs from cardiac progenitor cells are the most efficient in promoting angiogenesis per amount of VEGF-A protein produced. Intravenous administration of luciferase mRNA shows that EVs could distribute translatable mRNA to different organs with the highest amounts of luciferase detected in the liver. Direct injections of VEGF-A mRNA (via EVs or LNPs) into mice heart result in locally produced VEGF-A protein without spillover to liver and circulation. In addition, EVs from cardiac progenitor cells cause minimal production of inflammatory cytokines in cardiac tissue compared with all other treatment types. Collectively, the data demonstrate that LNPs transform EVs as functional extensions to distribute therapeutic mRNA between cells, where EVs deliver this mRNA differently than LNPs. 
  •  
12.
  • Nguyen, Duong T., et al. (author)
  • Humanizing Miniature Hearts through 4-Flow Cannulation Perfusion Decellularization and Recellularization
  • 2018
  • In: Scientific Reports. - : Nature Publishing Group. - 2045-2322. ; 8
  • Journal article (peer-reviewed)abstract
    • Despite improvements in pre-clinical drug testing models, predictability of clinical outcomes continues to be inadequate and costly due to poor evidence of drug metabolism. Humanized miniature organs integrating decellularized rodent organs with tissue specific cells are translational models that can provide further physiological understanding and evidence. Here, we evaluated 4-Flow cannulated rat hearts as the fundamental humanized organ model for cardiovascular drug validation. Results show clearance of cellular components in all chambers in 4-Flow hearts with efficient perfusion into both coronary arteries and cardiac veins. Furthermore, material characterization depicts preserved organization and content of important matrix proteins such as collagens, laminin, and elastin. With access to the complete vascular network, different human cell types were delivered to show spatial distribution and integration into the matrix under perfusion for up to three weeks. The feature of 4-Flow cannulation is the preservation of whole heart conformity enabling ventricular pacing via the pulmonary vein as demonstrated by noninvasive monitoring with fluid pressure and ultrasound imaging. Consequently, 4-Flow hearts surmounting organ mimicry challenges with intact complexity in vasculature and mechanical compliance of the whole organ providing an ideal platform for improving pre-clinical drug validation in addition to understanding cardiovascular diseases.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-12 of 12
Type of publication
journal article (11)
doctoral thesis (1)
Type of content
peer-reviewed (11)
other academic/artistic (1)
Author/Editor
Synnergren, Jane (5)
Risberg, Bo, 1941 (5)
Helenius, Gisela, 19 ... (4)
Johansson, Markus (3)
Jeppsson, Anders, 19 ... (3)
Sartipy, Peter (3)
show more...
Tangruksa, Benyapa (3)
Johansson, Bengt R, ... (2)
Valadi, Hadi, 1963 (2)
Esguerra, Maricris, ... (2)
Ulfenborg, Benjamin, ... (2)
Jennbacken, Karin (2)
Andersson, C. X. (2)
Kull, Bengt (2)
Siegbahn, Agneta (1)
Wiseman, John (1)
Nawaz, Muhammad (1)
Nelander, S (1)
Jing, Yujia (1)
Liu, Kai (1)
Lindfors, Lennart, 1 ... (1)
Althage, Magnus (1)
Hultin, Leif (1)
Camponeschi, Alessan ... (1)
Grimsholm, Ola, 1979 (1)
Christoffersson, Jon ... (1)
Carracedo, Miguel (1)
Nguyen, P (1)
Reyahi, Azadeh (1)
Li, Y. S. (1)
Fink, Helen, 1978 (1)
Miliotis, Tasso (1)
Albery, Tamsin (1)
Sundqvist, Martina (1)
Marco, Maugeri, 1983 (1)
Hicks, Ryan (1)
Mattsson, Erney, 195 ... (1)
Gan, Li Ming (1)
Yuan, S (1)
Li, Y. -C (1)
Davidsson, Pia (1)
Åvall, Karin (1)
Dekker, Niek (1)
González-King, Herná ... (1)
Rodrigues, Patricia ... (1)
Gurrapu, Ramya (1)
Silva, Andreia M. (1)
Musa, Gentian (1)
Kardasz, Dominika (1)
Rydén-Markinhuhta, K ... (1)
show less...
University
University of Gothenburg (11)
University of Skövde (6)
Uppsala University (1)
Chalmers University of Technology (1)
Karolinska Institutet (1)
Language
English (12)
Research subject (UKÄ/SCB)
Natural sciences (6)
Medical and Health Sciences (6)

Year

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Close

Copy and save the link in order to return to this view